Your activity: 20 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Acute myeloid leukemia in children and adolescents

Acute myeloid leukemia in children and adolescents
Authors:
Katherine Tarlock, MD
Todd M Cooper, DO
Section Editor:
Julie R Park, MD
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Dec 2022. | This topic last updated: Apr 21, 2021.

INTRODUCTION — Acute leukemia accounts for approximately 30 percent of all childhood malignancies and is the most common cancer in children. Acute myeloid leukemia (AML) accounts for approximately 15 percent of childhood leukemia and is much less common in the pediatric population than acute lymphoblastic leukemia (ALL), which accounts for 80 percent of pediatric acute leukemia. Survival rates for AML have greatly improved over the past several decades; however, overall survival for children with AML is approximately 65 to 70 percent and remains lower than for children with ALL [1]. The improvements in survival have been achieved through clinical trials investigating the role of intensification of therapy, including the use of allogeneic hematopoietic cell transplantation (HCT), as well as improvements in supportive care.

This topic will provide an overview of AML in children and adolescents, focusing on issues that are of interest to primary care providers. The pathogenesis of AML and discussions of the molecular genetics and cytogenetics in AML are presented separately. Transient myeloproliferative disorder of Down syndrome is also discussed separately. (See "Pathogenesis of acute myeloid leukemia" and "Molecular genetics of acute myeloid leukemia" and "Cytogenetic abnormalities in acute myeloid leukemia" and "Transient abnormal myelopoiesis (TAM) of Down syndrome (DS)".)

SPECIAL CONSIDERATIONS DURING THE COVID-19 PANDEMIC — The coronavirus disease 2019 (COVID-19) pandemic has increased the complexity of cancer care. Important issues include balancing the risk from treatment delay versus harm from COVID-19, ways to minimize negative impacts of social distancing during care delivery, and appropriately and fairly allocating limited health care resources. These issues and recommendations for cancer care during the COVID-19 pandemic are discussed separately.

(See "COVID-19: Considerations in patients with cancer".)

DIAGNOSIS

Clinical presentation — The most common presenting symptoms of AML are reflective of the leukemic burden. Similar to those with acute lymphoblastic leukemia (ALL), patients with AML can present with fever, malaise, musculoskeletal pains, lymphadenopathy, hepatosplenomegaly, and bleeding. A complete blood count most often reveals anemia and thrombocytopenia and can have decreased, normal, or increased white blood cell (WBC) counts with leukemic myeloblasts noted on the peripheral smear. The less common complications described below may require immediate medical intervention.

Disseminated intravascular coagulation (DIC) can be present and can range from mild to severe, especially in some subtypes of AML (eg, acute promyelocytic leukemia). Complications due to the leukemic burden at diagnosis may also include an elevated WBC of >100,000/microL, leading to leukostasis. (See 'Supportive care' below and 'Acute promyelocytic leukemia' below and "Overview of common presenting signs and symptoms of childhood cancer".)

Less often, children may present with symptoms of central nervous system (CNS) involvement (eg, headache, lethargy, mental status changes, cranial nerve palsies) or other extramedullary sites. Significant electrolyte derangements and acute kidney injury can occur, especially in those with high WBC or tumor burden. Hepatic dysfunction can also be present at diagnosis. (See 'Supportive care' below.)

Diagnostic evaluation — In addition to a history and physical examination, it is our practice to perform the following studies in children with suspected AML:

Laboratory studies – Laboratory studies include a complete blood count with differential, chemistries with liver and renal function and electrolytes, glucose, prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen, lactate dehydrogenase (LDH), calcium, magnesium, phosphorus, uric acid, albumin and total protein.

Lumbar puncture – Lumbar puncture for evaluation of the CNS. Cerebrospinal fluid (CSF) should be sent for cell count, protein, glucose, cytology (examination of stained cytospin slides). (See "Lumbar puncture: Indications, contraindications, technique, and complications in children".)

Bone marrow aspirate and biopsy – Bone marrow examination enables morphologic, immunophenotypic, cytogenetic (ie, karyotype and fluorescence in situ hybridization [FISH]), and molecular testing, which are essential for accurate risk stratification [2]. In some settings, immunophenotypic evaluation by flow cytometry of the peripheral blood can be used for diagnostic purposes, but this does not replace bone marrow evaluation.

Extramedullary disease – Patients with suspected extramedullary disease should have appropriate radiographic imaging of the suspected sites.

Screen for familial AML – Screening for familial acute leukemia and myelodysplastic syndromes consists of a careful medical and family history aimed at identifying signs and symptoms of the known familial syndromes. This is discussed in more detail separately. (See "Familial disorders of acute leukemia and myelodysplastic syndromes", section on 'Evaluation of patients with AL or MDS'.)

Pathologic features — The pathologic evaluation of suspected AML includes an assessment of blast morphology, immunophenotype, cytogenetics, and molecular features. AML is distinguished from ALL based on the morphology and immunophenotype of the blasts. Findings on metaphase cytogenetics, FISH, and molecular studies are important for risk stratification. Lumbar puncture with CSF analysis is a standard part of the evaluation of children with newly diagnosed AML. The pathologic features of AML are presented in more detail separately. (See "Clinical manifestations, pathologic features, and diagnosis of acute myeloid leukemia", section on 'Pathologic features'.)

Morphology – Myeloblasts are immature cells with large nuclei, usually with prominent nucleoli, and a variable amount of pale blue cytoplasm. Typically, myeloid blasts have more abundant cytoplasm compared with lymphoid blasts and may have Auer rods or granules present (picture 1). The morphologic classification of AML has been based upon the French-American-British (FAB) classification that relies on the lineage associated phenotype, ranging from minimally differentiated (FAB M0) to the more mature acute megakaryoblastic leukemia (AMKL, FAB M7). (See "Classification of acute myeloid leukemia (AML)", section on 'AML not otherwise specified'.)

Immunophenotype – The immunophenotype of the tumor cells can be determined by flow cytometry or by immunohistochemistry. The tumor cell immunophenotype can rapidly distinguish between AML and ALL. It can also determine myeloid-specific lineage but usually does not substitute for morphologic FAB classification. AML expresses numerous markers, including CD11b, CD34, CD33, CD45, CD64, CD65, CD117, myeloperoxidase (MPO), and lysozyme.

Cytogenetics – Cytogenetic abnormalities can be detected in approximately 75 percent of pediatric AML, and several findings have prognostic or therapeutic implications [3]. Many abnormalities can be detected by conventional karyotype, but some translocations can be cryptic and require FISH or DNA sequencing for detection or confirmation. In addition to conventional karyotyping, FISH should be performed to evaluate for t(8;21), inv(16), t(15;17), and 11q23 translocations. (See "Cytogenetic abnormalities in acute myeloid leukemia" and "Molecular genetics of acute myeloid leukemia".)

Mutation analysis Pediatric AML is genetically heterogeneous, the molecular profiles differ from adult AML, and certain mutations or genetic profiles have prognostic or therapeutic implications [4,5]. We suggest mutation analysis with polymerase chain reaction (PCR) and/or next-generation sequencing (NGS). Testing should include NPM1, CEBPA, FLT3/ITD, KIT, and WT1. Comprehensive sequencing that utilizes DNA and RNA sequencing can be utilized to define the genomic profile, including cryptic fusions not detected by conventional cytogenetics [4]. (See "Molecular genetics of acute myeloid leukemia", section on 'Gene mutations'.)

CSF analysis – The diagnosis of CNS involvement requires either cytologic confirmation of leukemia cells in the CSF, clinical signs of CNS leukemia (eg, facial nerve palsy, brain/eye involvement), or a tumor mass detected by imaging. CNS positivity based on cytology is generally defined as >5 x106/L WBCs in the CSF with the presence of blasts in a non-bloody lumbar puncture.

RISK STRATIFICATION

Overview — Risk stratification in pediatric AML is based on cytogenetic and molecular features, including genomic profiling, and response to induction therapy. While there is general agreement on low-risk cytogenetic features, there is controversy about certain high-risk molecular features because some cooperative groups use different definitions for treatment response.

We suggest use the following risk stratification scheme, which takes into account cytogenetic and molecular features and response to induction chemotherapy:

Favorable:

t(8;21)(q22;q22); RUNX1-RUNX1T1

inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11

Mutated NPM1 without FLT3-ITD (normal karyotype)

Mutated CEBPA (normal karyotype)

Intermediate – Cases that do not harbor cytogenetic or molecular abnormalities that classify them as favorable or adverse risk are categorized as intermediate risk. This population is further subclassified based on response to induction therapy. (See 'Response to induction chemotherapy' below.)

Adverse:

t(6;9)(p23;q34)/DEK-NUP214

NUP98-NSD1

Certain KMT2A rearrangements (see 'High-risk cytogenetics' below)

CBFA2T3A-GLIS2

Abnormalities of 3q

Monosomy 5 or del(5q)

Monosomy 7

Complex karyotype

High allelic ratio FLT3-ITD

Residual disease following induction therapy (see 'Response to induction chemotherapy' below)

Details regarding the impact of each of these findings are presented in the following sections. There are less data regarding rare subtypes, such as non-Down syndrome acute megakaryoblastic leukemia (AMKL), although results from comprehensive genomic sequencing allows for a more refined risk stratification in this setting [4].

Cytogenetics

Favorable cytogenetics — AML with favorable cytogenetics accounts for approximately 20 to 30 percent of pediatric AML and includes those who harbor the chromosomal abnormalities t(8;21) and inv(16) or t(16;16), also called core binding factor (CBF) AML [5,6]. Treatment on contemporary protocols with chemotherapy alone results in a relapse-free survival rate of approximately 70 percent and an overall survival of approximately 80 percent [6-8]. For patients with non-Down syndrome acute megakaryoblastic leukemia (AMKL), those harboring the RBM15-MKL1 translocation have a more favorable prognosis with an overall survival of approximately 70 percent [4]. (See "Cytogenetic abnormalities in acute myeloid leukemia", section on 't(8;21); RUNX1-RUNX1T1' and "Cytogenetic abnormalities in acute myeloid leukemia", section on 'inv(16) or t(16;16); CBFB-MYH11'.)

The hallmark diagnostic feature of acute promyelocytic leukemia (APL) is the balanced translocation between the PML gene on chromosome 15 and RARA (RAR-alpha) gene on chromosome 17 resulting in the t(15;17)(q22;q21) [9]. Patients with APL can achieve excellent outcomes when treated with appropriate regimens. (See 'Acute promyelocytic leukemia' below and "Molecular biology of acute promyelocytic leukemia".)

High-risk cytogenetics — Children with adverse cytogenetic features account for approximately 15 percent of pediatric AML [6]. Patients with poor-risk cytogenetics include those that lack any favorable changes and harbor any of the following cytogenetic abnormalities: monosomy 7, monosomy 5, deletion of 5q, abnormalities of 3q, t(6;9)(p23;q34), and complex karyotype (defined as three or more cytogenetic abnormalities) [5,10,11]. Children and adolescents harboring these unfavorable features have long-term survival <50 percent, and in some cases <20 percent [5].

For patients with non-Down syndrome acute megakaryoblastic leukemia (AMKL), the cytogenetic abnormalities CBFA2T3-GLIS2, NUP98-KDM5A, and MLL (KMT2A) rearrangements have a poor prognosis compared to patients with normal karyotypes as well as other cytogenetic lesions, and experience an long-term survival of approximately 30 to 40 percent [4]. (See "Prognosis of acute myeloid leukemia", section on 'Karyotype'.)

Translocations involving KMT2A (previously referred to as MLL) at the 11q23 locus are present in 15 to 20 percent of pediatric AML cases. There are many fusion partners of KMT2A and prognosis may vary according to the fusion partner. Rearrangements including t(6;11)(q27;q23), t(10;11)(p11.2;q23.3) and t(10;11)(p12;q23.3) have been associated with a poor prognosis [12,13].

Abnormalities involving 3q, including inv(3)(q21q26.2) and t(3;3)(q21;q26.2), (RPN1-EVI1) are associated with adverse prognosis in pediatric AML [5,13]. In addition, NUP98-NSD1 fusions, which commonly occur with FLT3-ITD mutations, are associated with a poor prognosis in children and adults, and are considered a high-risk lesion [13-16].

Other notable cytogenetic abnormalities — Rearrangements involving KMT2A are most prevalent in young children, especially those <2 years [17]. There are many fusion partners of KMT2A and prognosis may vary according to the fusion partner. The most common rearrangement in pediatric AML involving KMT2A is t(9;11)(p22;q23), which accounts for approximately 50 percent of cases, but this is not associated with adverse outcomes [12]. The t(1;11)(q21;23) rearrangement has been associated with a more favorable outcome relative to other KMT2A fusions [12,13].

Trisomies are common events in pediatric AML, often involving chromosome 8, 19, and 21 and can occur alone or in conjunction with additional karyotypic abnormalities.

Molecular features — Several recurring mutations have been identified in pediatric AML, some of which are predictive of relapse and prognosis. Testing for these mutations is now an integral part of the diagnostic workup and subsequent risk stratification.

High-risk molecular features — Activating mutations in the FLT3 gene are among the most common recurring somatic mutations in AML. Of these, internal tandem duplications of FLT3 (FLT3/ITD) are the most frequent and occur in approximately 10 to 15 percent of pediatric AML [18,19]. The prevalence of FLT3/ITD increases with age, occurring in 15 to 25 percent of adolescents and young adults. Activating loop mutations of FLT3 (FLT3/ALM) that result from point mutations occur at a lower prevalence of approximately 5 to 7 percent and have no prognostic impact [20]. (See "Molecular genetics of acute myeloid leukemia", section on 'FLT3'.)

Patients with mutations involving FLT3/ITD with a high allelic ratio (HAR) of mutant to wild-type FLT3 have a poor prognosis. Although the cutoff used differs by cooperative group, it is generally accepted that an allelic ratio greater than 0.4 to 0.5 is considered HAR and a marker of poor prognosis [20,21]. Patients with HAR FLT3/ITD have a significantly higher risk of being refractory to induction chemotherapy as well as risk of relapse [18]. Children with HAR FLT3/ITD mutations have overall survival rates of approximately 20 to 30 percent with contemporary chemotherapy regimens alone; however, survival improves to approximately 50 to 60 percent when consolidation allogeneic hematopoietic cell transplantation (HCT) is employed [20,21]. (See 'Post-remission therapy' below.)

FLT3 mutations provide a therapeutic target for novel agents. (See 'FLT3 inhibitors' below.)

Low-risk molecular features — In pediatric AML, a low-risk molecular profile includes cases with mutations involving CEBPA and those with NPM1 mutations in the absence of FLT3/ITD:

Mutations involving CEBPA occur in approximately 5 percent of pediatric AML. Children with CEBPA mutations have a more favorable prognosis with overall survival rates of approximately 70 percent following treatment with chemotherapy alone [22]. (See "Prognosis of acute myeloid leukemia", section on 'CEBPA gene'.)

Mutations involving NPM1 are present in approximately 8 to 10 percent of pediatric AML. NPM1 mutations often co-occur with FLT3/ITD mutations, so it is important to evaluate for additional mutations [23]. Children with NPM1 mutations that do not harbor the unfavorable FLT3/ITD demonstrate a more favorable prognosis with an estimated survival rate of approximately 70 percent with chemotherapy alone [23]. (See "Prognosis of acute myeloid leukemia", section on 'NPM gene' and "Molecular genetics of acute myeloid leukemia", section on 'NPM1 mutations'.)

Other molecular features — Other recurring mutations, including WT1, KIT, NRAS, and KRAS, have been identified in pediatric AML. Their prognostic significance has not been determined, but their non-random association with additional cytogenetic and molecular aberrations serves as evidence that they likely serve as cooperating mutations in leukemogenesis [24-27]. Additional information about the impact of these gene mutations in adult AML is presented separately. (See "Molecular genetics of acute myeloid leukemia", section on 'Gene mutations' and "Prognosis of acute myeloid leukemia", section on 'Gene mutations'.)

Emerging technologies such as next generation sequencing provide further insights into mechanisms of leukemogenesis; findings include detection of cryptic fusion genes, copy number alternations (CNA; focal gains or losses of genomic regions), loss of heterozygosity (LOH), and distinct immunophenotypes [28-30]. Data regarding these genomic alterations may provide additional prognostic information in pediatric AML and could suggest potential therapeutic strategies in pediatric AML.

Further examination of the clinical impact of these genetic features will be critical to their incorporation into upfront risk stratification and therapeutic allocation.

Response to induction chemotherapy — A significant number of patients do not harbor cytogenetic or molecular abnormalities with prognostic value, and are thus classified as intermediate risk. For this group of patients, the most important prognostic factor is the disease response to induction therapy, including an assessment of measurable residual disease (MRD; also referred to as minimal residual disease) by flow cytometry [31-33]:

MRD negative – Children in this intermediate-risk group who achieve a remission following the first course of induction (as defined by the absence of MRD detected by flow cytometry) experience a more favorable relapse-free survival of approximately 65 percent [32].

MRD positive – Children with detectable disease by flow cytometry following induction experience a significantly worse prognosis with a relapse risk of approximately 60 percent [32].

Among children with favorable or unfavorable cytogenetic or molecular features, the role of MRD at the end of induction has not yet been shown to have definitive prognostic significance [32,34]. Detection of MRD by additional methods, such as expression of leukemia-associated genes or disease-specific mutations by polymerase chain reaction (PCR), is somewhat more limited in its use as compared with flow cytometry but can also provide sensitive methods to detect low level of residual leukemia and can inform prognostic and therapeutic decisions [35,36]. (See "Induction therapy for acute myeloid leukemia in medically-fit adults", section on 'Remission assessment'.)

TREATMENT — Chemotherapeutic regimens in children and adolescents most often contain two courses of intensive induction chemotherapy followed by either cytarabine-based consolidation or hematopoietic cell transplantation (HCT). Children and adolescents with AML should be treated in the context of a clinical trial whenever possible. Trials are designed to compare potentially better therapy with that which is currently accepted as standard. Additional information and instructions for referring a patient to an appropriate research center can be obtained from the United States National Institutes of Health.

Induction — Standard induction chemotherapy consists of an anthracycline (eg, daunorubicin, idarubicin, or the anthracenedione mitoxantrone) and extended exposure to cytarabine. For children with newly-diagnosed therapy-related AML or AML with myelodysplasia-related changes, the US Food and Drug Administration (FDA) approved CPX-351, a liposomal combination of daunorubicin and cytarabine, for children ≥1 year [37]. In some regimens a third drug, such as etoposide or 6-thioguanine, may be included, but the benefit of these additional agents has not yet been proven [38].

In a phase 3 trial for pediatric AML, the antibody-drug conjugate gemtuzumab ozogamicin (GO; which targets CD33), decreased rates of relapse compared to chemotherapy only [7]. GO is approved by the US Food and Drug Administration for the treatment of newly diagnosed AML in adults and for treatment of relapsed or refractory CD33-positive AML in adults and in pediatric patients one month and older. When administered as part of induction chemotherapy, GO is generally given as single dose during the first week of therapy and achieve complete remission (CR) rates >70 percent [1,38].

Post-remission therapy — Consolidation with high dose chemotherapy or HCT may be administered after achieving remission. Consolidation chemotherapy using high-dose cytarabine (HiDAC) reduced the rate of relapse in several clinical trials in children [39-41]. The decision to proceed with allogeneic HCT as consolidation must weigh the risk of relapse with the increased risk of mortality and morbidity associated with HCT. Allogeneic HCT is offered to children with high-risk cytogenetics in first CR. Patients with favorable-risk disease features, such as t(8;21) and inv(16), should not be transplanted in first CR. Children with therapy-related AML (ie, after treatment with cytotoxic chemotherapy or radiation therapy for another cancer) have adverse outcomes and should receive consolidation HCT [42].

Both autologous and allogeneic HCT have been investigated in pediatric AML as a strategy to improve survival. No benefit has been demonstrated for autologous HCT compared with nonmyeloablative chemotherapy for children with AML who have achieved remission [43-45]. The role of allogeneic HCT in children and adolescents with AML continues to be an area of active investigation and there is no general consensus among experts. When compared with chemotherapy alone, post-remission allogeneic HCT has been associated with a significantly lower relapse risk in some studies [2,39,46]. However, when applied to all patients, this has not always resulted in improved survival. This discrepancy may be due to the higher treatment-related mortality of allogeneic HCT.

It is generally accepted to offer patients with poor-risk cytogenetics allogeneic HCT as consolidation therapy, although the data supporting this approach are limited. Even with HCT, these children continue to fare poorly. Many studies of patients with poor-risk cytogenetics show no survival benefit using HCT as consolidation therapy compared with chemotherapy alone [47,48]. However, there are published data demonstrating the benefit of HCT for some patients with high-risk disease features in first CR [47,49-51]. As an example, allogeneic HCT in first CR improves survival in children with high allelic burden of FLT3/ITD [21,52,53].

In general, matched sibling donors are a preferred donor source when available; however, unrelated donor sources can yield equivalent outcomes [54]. (See "Donor selection for hematopoietic cell transplantation".)

FLT3 inhibitors — The FLT3-ITD mutation is amenable to targeted agents. The RATIFY trial in adults with FLT3 mutations demonstrated the addition of the FLT3 inhibitor midostaurin to conventional chemotherapy was safe and improved outcomes for patients with FLT3-ITD mutations [55], but its safety and efficacy have not been demonstrated in children. The FLT3 inhibitor sorafenib was associated with decreased relapse and improved survival in children when compared to historical controls not treated with sorafenib [56]. Continued therapy with a FLT3 inhibitor for up to two years after HCT is associated with superior outcomes in adults, and this strategy has also shown to be safe in children [57-60].

Midostaurin is not approved by the US FDA for treatment of children with FLT3 mutated AML.

CNS-directed therapy — Treatment of the central nervous system (CNS) with intrathecal chemotherapy should be given to all children and adolescents with AML, including those without detectable CNS involvement at diagnosis. The intrathecal therapy used varies according to cooperative group protocol. Single agent cytarabine is commonly used, but some protocols use triple therapy consisting of cytarabine, methotrexate, and hydrocortisone. Although cranial irradiation is effective at preventing CNS leukemia, it should not be used prophylactically due to the risks of late effects including cognitive deficits, endocrine deficits, and secondary malignancies [61-63]. The use of HiDAC as systemic therapy, which crosses the blood brain barrier, provides additional CNS-directed therapy. The optimal number of prophylactic intrathecal treatments remains unknown, most regimens and clinical trials provide at least one dose with each course of therapy.

CNS involvement occurs in 5 to 10 percent of children with AML at diagnosis [64]. Risk factors associated with CNS involvement include age <2 years, high white blood cell (WBC) count, hepatosplenomegaly, t(8;21), inv(16), or t(16;16) [64,65]. Patients with CNS involvement require augmented CNS-directed therapy. CNS positivity at diagnosis is not an adverse prognostic factor for children with AML, and with intensified CNS-directed therapy does not affect overall survival [64]. Frequent intrathecal chemotherapy combined with intensive systemic chemotherapy, including HiDAC, may be as effective as cranial irradiation in patients with CNS involvement and radiation may not be necessary [64,65].

Myeloid sarcomas — Myeloid sarcoma (MS), also known as chloromas, refers to deposits of myeloid blasts outside the bone marrow that may cause destruction or compression in normal tissue. Sites of MS most commonly include the CNS, skin, orbit, and bone. The incidence of MS in pediatric AML is approximately 10 percent and is more common in patients with the following features [66,67]:

Younger age

High white blood cell count at diagnosis

t(8;21)

French-American-British (FAB) M4 and M5 morphology

Some studies in adults have demonstrated high relapse rates and lower survival in patients with MS. In children, the prognostic impact of MS appears to depend upon the site of involvement [64,66,67]. Pediatric patients with MS involving the CNS or orbit have demonstrated improved survival compared with those without CNS/orbit involvement and patients without MS. In contrast, children with chloromatous skin involvement have higher rates of relapse. Extramedullary relapses are more common among those with extramedullary disease at diagnosis. Focal radiation may provide symptomatic benefit, however, it does not appear to offer any additional progression-free or overall survival benefit to chemotherapy in patients with MS [66,68].

Acute promyelocytic leukemia — Acute promyelocytic leukemia (APL) is a rare subtype of AML that is characterized by maturation arrest at the promyelocyte stage and the classic chromosomal translocation t(15;17)(q22;q21) [9]. (See "Molecular biology of acute promyelocytic leukemia".)

APL is risk stratified based on the presenting WBC count, with patients presenting with a WBC <10x109/L (<10,000/microL) classified as standard risk and those with a WBC >10 x 109/L classified as high risk [69]. Patients with high WBC counts experience higher rates of relapse and are especially susceptible to early mortality due to coagulopathy (ie, disseminated intravascular coagulation [DIC]). Many patients with APL present with DIC that requires urgent and aggressive management. All-trans retinoic acid (ATRA) should be started quickly when APL is suspected [70].

ATRA, which targets the PML-RARA fusion protein and induces differentiation of the blasts, is a critical component of successful treatment of APL [71]. Anthracyclines have been used historically as a mainstay of treatment in APL, and when combined with ATRA and arsenic trioxide (ATO), result in excellent survival [72-75]. Clinical trials in adults have demonstrated equivalent survival in standard-risk patients when treated with ATRA and ATO alone, while anthracycline is utilized in induction for high-risk patients [76,77]. Retrospective studies in children have shown a regimen of ATRA plus ATO for standard risk APL is well tolerated and results in excellent outcomes, similar to adults [78]. The management of APL in adults is presented in detail separately. (See "Initial treatment of acute promyelocytic leukemia in adults".)

DOWN SYNDROME AND AML — Down syndrome (DS) is well recognized as a cancer predisposition syndrome. Children with DS have 10 to 20 times greater risk of developing AML compared with children without DS. Infants with DS can also develop the transient myeloproliferative disorder (TMD) of DS, also known as transient abnormal myelopoiesis (TAM), which occurs at a prevalence of approximately 10 percent. TMD is a clonal myeloproliferative disorder that presents with peripheral leukocytosis and the appearance of megakaryoblasts in the peripheral blood that are identical to those seen in acute megakaryoblastic leukemia (AMKL). Complications include pericardial and pleural effusions, coagulopathy, hydrops fetalis, hepatic dysfunction, and hepatic fibrosis [79-81]. TMD is discussed in more detail separately. (See "Transient abnormal myelopoiesis (TAM) of Down syndrome (DS)".)

For the majority of infants of who develop TMD, the disease resolves spontaneously without any treatment and over 50 percent of infants experience resolution by six months of age [81]. Approximately 20 to 25 percent of infants with TMD require treatment with chemotherapy, which is often indicated due to respiratory compromise, liver dysfunction, or hyperviscosity associated with hyperleukocytosis [81]. Infants who are diagnosed with TMD are followed with frequent complete blood counts with differential in the first five years of life as approximately 20 to 25 percent of infants can develop AML, even after the resolution of TMD [7,79]. Children with longer time to resolution of TAM may have a higher risk of subsequent AML, and most cases of AML in children with DS present by four years of age [81].

AML in children with DS has unique biologic characteristics that greatly influence therapy and prognosis. Children with DS commonly develop AMKL, a subtype of AML that is rare in children without DS [82,83]. The additional chromosome 21 is recognized as important factor in leukemia predisposition. A key event in the leukemogenesis of DS-AML, as well as TMD, is the acquisition of mutations in the GATA1 gene, which is important in hematopoietic development [84,85].

Overall, children with DS have superior overall survival when compared with children with AML who do not have DS. Leukemic cells of DS-AML exhibit increased sensitivity to cytarabine and daunorubicin [86,87], and children with DS-AML can experience excellent outcomes without the intensive therapy that is needed to achieve cure in pediatric AML [38,88-91]. A multicenter trial of 204 patients with DS-AML who received high dose cytarabine early in the course of treatment and a reduced cumulative dose of daunorubicin resulted in five-year event free survival and overall survival of 90 and 93 percent, respectively [92].

Importantly, children with DS have higher rates complications from intensive chemotherapy, namely infection and cardiotoxicity [83,93]. It is essential that children with DS who develop AML are treated with DS-specific therapy as they cannot tolerate the toxic effects of the intensive pediatric AML regimens. Chemotherapy regimens using a backbone of cytarabine and an anthracycline are effective at achieving remission and can cure the majority of patients, especially when toxicity is minimized [88,94,95]. A multicenter trial of 170 children with DS-AML demonstrated that reduction of cumulative etoposide, less intrathecal CNS prophylaxis, and elimination of maintenance therapy did not impact survival with five-year event-free survival of 87 percent and overall survival of 89 percent [96]. Modern clinical trials have focused on increased utilization of cytarabine, lower doses of anthracyclines, as well as enhanced supportive care.

Unfortunately, children with DS who achieve inadequate response or relapse following therapy have very poor outcomes and are not good candidates for conventional intensity allogeneic hematopoietic cell transplantation (HCT) [97]. Children with DS who receive HCT for AML have very poor outcomes that are attributed to high treatment-related morbidity and incidence of relapse [98].

SUPPORTIVE CARE — Improvements in supportive care have been essential to the improved outcomes observed in pediatric AML over the past several decades. Children and adolescents with AML are at risk for significant morbidity and mortality both from their disease and the direct effects of curative therapy.

Hyperleukocytosis — Hyperleukocytosis is defined as a white blood cell (WBC) count of >100,000/microL (>100 x 109/L) and is a risk factor for significant complications including leukostasis, disseminated intravascular coagulation (DIC), and tumor lysis syndrome (TLS). The risk of serious complications and mortality from these issues is highest at the time of diagnosis and within the days following the initiation of therapy [99]. It is important to recognize that the WBC count is a somewhat arbitrary threshold and does not always predict the presence of complications. Compared with acute lymphoblastic leukemia (ALL), complications of hyperleukocytosis are more common and can occur at comparatively lower WBC counts in AML [100]. In pediatric AML, higher WBC counts are more common in patients presenting with FLT3/ITD, MLL rearrangements, FAB M4 and M5 phenotypes, and the CBF lesions [20,99,101].

Leukostasis – Hyperleukocytosis may lead to tissue damage from blast infiltration and leukostasis, resulting in hemorrhagic and thromboembolic events. The clinical presentation of leukostasis varies depending on the affected organs. Patients with lung involvement may present with dyspnea, hypoxemia, or respiratory failure while neurologic involvement may manifest as somnolence, coma, or focal neurologic deficits. Leukostasis is an oncologic emergency and can be life threatening, especially the pulmonary and neurologic complications [102]. Although leukapheresis or exchange transfusion can effectively lower the WBC count temporarily, these procedures pose significant risk, are temporary, and can exacerbate thrombocytopenia and risk of DIC [103]. Due to the risks and limitations of these procedures, they are not generally recommended for pediatric AML patients without symptoms of leukostasis [104,105]. The most effective means at achieving reduction in WBC is prompt diagnosis and initiation of chemotherapy [102].

Tumor lysis syndrome – Supportive management should include hyperhydration to reduce risk of tumor lysis and decrease blood viscosity. TLS may manifest as hyperphosphatemia, hypocalcemia (caused by precipitation of calcium phosphate), hyperuricemia, hyperkalemia, and acute renal failure. Rapid leukemic cell lysis after chemotherapy can cause over-production and over-excretion of uric acid. The precipitation of uric acid in the tubules can lead to oliguric or anuric renal failure known as uric acid nephropathy. Administration of prophylactic medications to prevent complications of TLS, such as allopurinol or rasburicase, and careful monitoring of renal function is essential. (See "Tumor lysis syndrome: Prevention and treatment".)

Disseminated intravascular coagulation – Acute DIC occurs most commonly in patients presenting with hyperleukocytosis, monocytic subtypes (FAB M5), or with acute promyelocytic leukemia (APL). If APL is suspected, all-trans retinoic acid (ATRA) should be started immediately before the diagnosis is confirmed as it is the most important treatment of DIC in this population. Supportive care of patients with AML-associated DIC should include standard management with appropriate blood products (eg, platelets and coagulation factors), which is essential to minimize bleeding risk. (See "Disseminated intravascular coagulation in infants and children".)

Infectious complications — The intensity of the treatment required to cure AML carries a significant risk of severe infections with bacteria, viruses, and fungi. The infectious risk is due, in part, to alterations in mucosal barriers and the prolonged absence of neutrophils. Adolescents and young adults may be at higher risk for treatment-related toxicity compared with younger children [106,107].

Prompt antibiotic administration is critical for any child with fever who is undergoing treatment for AML; management of fever in this setting is discussed separately. (See "Fever in children with chemotherapy-induced neutropenia".)

Bacterial infections — Bacteremia is a leading cause of morbidity and mortality in children who are treated with intensive chemotherapy for AML, and viridans group streptococci and gram-negative bacteria are common pathogens in this setting. Antibiotic prophylaxis can reduce the risk of bacteremia during intensive chemotherapy treatment, but there is no demonstrated role for prophylactic administration of granulocyte colony-stimulating factor (G-CSF) [108-110].

The role of antibiotic prophylaxis in children undergoing AML remission induction therapy is controversial. While we do not routinely treat with antibiotic prophylaxis, some experts do suggest prophylactic quinolones. The reasoning behind our approach is that although levofloxacin prophylaxis can decrease the incidence of bacteremia and febrile neutropenia in this setting, it has not been shown to reduce sepsis or improve survival; the benefits must be balanced against the potential for toxicity and/or complications, and should be considered from the perspective of institutional resistance patterns and the potential for emergence of antibiotic resistance. We favor more data to support a benefit for critical outcomes such as sepsis or survival before routinely suggesting this approach. However, it is worth noting that fluoroquinolone prophylaxis is suggested for adults who are at high risk for neutropenic fever. (See "Prophylaxis of infection during chemotherapy-induced neutropenia in high-risk adults", section on 'Indications'.)

A multicenter trial randomly assigned 195 children (age 6 months to 21 years) who were undergoing two cycles of intensive chemotherapy for AML or relapsed ALL to levofloxacin prophylaxis versus no prophylaxis [108]. Compared with no prophylaxis, the likelihood of bacteremia in children receiving levofloxacin prophylaxis was reduced (21.9 versus 43.4 percent, respectively; risk difference, 21.6 percent; 95% CI 8.8-34.4 percent), and the study was terminated early when a planned interim analysis demonstrated the efficacy of levofloxacin. Levofloxacin prophylaxis was also associated with fewer episodes of fever and neutropenia (71.2 versus 82.1 percent), but there were no differences between the levofloxacin and placebo groups in the other secondary outcomes (ie, severe infections, invasive fungal disease, Clostridioides [formerly Clostridium] difficile-associated diarrhea, musculoskeletal toxic effects). Notably, the reduction of bacteremia for children with AML was modest (23.4 versus 29.7 percent), and there was no benefit in a cohort of children undergoing allogeneic hematopoietic cell transplantation (HCT); the effect was most pronounced in children with relapsed ALL. Viridans group streptococci were the most commonly cultured organisms, and in the small sample of isolates available for sensitivity testing, all five isolates from children receiving prophylaxis were resistant to levofloxacin, whereas all eight isolates from the untreated patients were sensitive.

Viridans group streptococci can cause severe sepsis and viridans streptococcal shock syndrome (SVS), resulting in hemodynamic instability and acute respiratory distress syndrome. The risk of infection with viridans group streptococci is more pronounced in patients treated with high dose cytarabine (HiDAC) and those with prolonged neutropenia [111]. Among children treated for AML, as many as 25 percent develop SVS [112]. Prompt recognition of the potential for SVS is critical as rapid progression can be fatal and appropriate intravenous antibiotics should be started immediately [113]. Details regarding the choice of antibiotic therapy are presented separately. (See "Fever in children with chemotherapy-induced neutropenia".)

Fungal infections — Severe fungal infections, including those caused by yeasts (eg, Candida spp) and invasive molds (eg, Aspergillus spp) pose a significant risk throughout AML therapy and for those undergoing allogeneic HCT [114]. Antifungal prophylaxis is effective at reducing invasive fungal infections and should be incorporated into management of children and adolescents receiving therapy for AML [110,115]. New or prolonged fevers in the setting of neutropenia should prompt suspicion of fungal infection, and empiric antifungal therapy should be administered immediately for presumed or documented fungal infections; treatment should not be delayed for completion of the diagnostic workup [116-118]. (See "Fever in children with chemotherapy-induced neutropenia".)

Antifungals used for prophylaxis include fluconazole, which has activity against Candida spp, [119] and voriconazole, posaconazole, and itraconazole, which have activity against both Candida spp and molds, such as Aspergillus spp [120-123]. The choice of regimen depends upon the most likely pathogen, and specific practices vary between centers. Consultation with Infectious Disease specialists should be considered in cases of presumed or documented infections. (See "Treatment and prevention of invasive aspergillosis".)

Initial evaluation of new or prolonged fevers in the setting of neutropenia should include computed tomography (CT) of the chest, abdomen, pelvis, and sinuses [124]. If a patient has additional symptoms that involve extremities or the central nervous system (CNS), appropriate imaging and diagnostic testing, including examination of the cerebrospinal fluid (CSF), should be obtained. Among patients with radiographic lung findings suspicious for fungal infection, additional diagnostic testing may include bronchial alveolar lavage or biopsy of suspicious lesions [118].

RELAPSED LEUKEMIA — Approximately 30 percent of children with AML will experience relapse. In general, the prognosis for these children is poor with about one-third of children being cured. Patients with core binding factor AML and other favorable molecular lesions are more likely to achieve long-term survival. Survival is especially poor for patients with high-risk disease features or those who have previously received allogeneic hematopoietic cell transplantation (HCT). The time to relapse is an important prognostic factor, and survival is less than 20 percent for patients relapsing within one year of achieving complete remission (CR) [125].

Re-induction regimens for children and adolescents with relapsed AML vary, but commonly include high-dose cytarabine (HiDAC) with or without an anthracycline. The use of an anthracycline depends on cardiac function at relapse and the cumulative anthracycline dose with initial therapy. Regimens often combine HiDAC with other agents such as fludarabine and clofarabine, and anthracyclines when appropriate [126-128]. Gemtuzumab ozogamicin (GO) has also shown responses in relapsed CD33 positive AML, either as a single agent or in combination with additional chemotherapy agents [129,130]. GO is approved by the US Food and Drug Administration for treatment of relapsed AML in children, as a single agent.

Achievement of a second CR is the most important prognostic factor in long-term survival, and patients who have no evidence of measurable residual disease (MRD; also referred to as minimal residual disease) following second-line therapy and prior to HCT have the best outcomes [126]. Consolidation HCT in CR is recommended for patients that relapse. Early phase clinical trials with novel agents should also be considered for all children and adolescents who relapse.

LATE EFFECTS — Advances in the treatment of childhood and adolescent AML have resulted in an increasing number of cancer survivors and have brought important attention to the late effects of chemotherapy and allogeneic HCT. Although information regarding late effects is not as robust as for acute lymphoblastic leukemia (ALL), potential late adverse effects include late onset cardiotoxicity, central nervous system (CNS) impairment, decreased linear growth, infertility, cataracts, and an increased incidence of secondary cancers as well as an overall decreased health status due to such factors as neurocognitive dysfunction, depression, fatigue, and anxiety. (See "Assuring quality of care for cancer survivors: The survivorship care plan" and "Overview of cancer survivorship in adolescents and young adults".)

Anthracyclines remain very effective agents in AML but carry a risk of early as well as late onset cardiotoxicity, which in the most severe cases results in heart failure. The cumulative dose of anthracyclines is the most important risk factor for late cardiac effects. Although there is no dose of anthracyclines that is recognized to be safe, cumulative anthracycline doses of >300 mg/m2 result in significantly higher rates of late cardiac compromise and this risk rises even more steeply in excess of 400 mg/m2 [131,132]. Current United States and European treatment protocols for AML utilize between 300 to 440 mg/m2 cumulative anthracycline dosing and estimated incidences of late cardiac effects are 3 to 5 percent [133,134]. Children and adolescents treated for AML should receive long-term follow-up with routine cardiac monitoring.

Survivors of childhood AML are at risk for secondary neoplasms, mainly solid tumors, and long-term follow-up data demonstrate a prevalence of approximately 1.7 percent [135]. This risk is increased in patients who receive radiation, which is often a part of HCT conditioning [136].

The intensity of therapy for AML, especially radiation therapy for HCT, also puts survivors at risk for endocrine dysfunction, with the thyroid and gonadal systems the most affected [135-137]. Children who receive total body irradiation or cranial irradiation are also at risk for growth defects and neurocognitive dysfunction; this is more pronounced in very young children who receive radiation [138].

The occurrence of specific complications depends on the patient's age and the type and intensity of therapy with which they were treated. Specific long-term follow-up guidelines after treatment of childhood cancer have been published by the Children's Oncology Group, and are available at www.survivorshipguidelines.org.

FUTURE DIRECTIONS — Children and adolescents with AML should be treated in the context of a clinical trial whenever possible. Trials are designed to compare potentially better therapy with that which is currently accepted as standard. Additional information and instructions for referring a patient to an appropriate research center can be obtained from the United States National Institutes of Health.

New technologies, such as next-generation sequencing, that can more deeply interrogate AML at the genomic and epigenomic level have the potential to identify novel biomarkers that can enhance risk stratification and potential therapeutic targets. To date, increased understanding of AML at the molecular level has provided insights into molecular lesions that can be targeted for therapy in pediatric AML. The FLT3/ITD mutation is one of the first genomic lesions in AML to be directly targeted with FLT3 inhibitors and demonstrated efficacy in adults and children with this mutation. As an example, the potent FLT3 inhibitor gilteritinib is active in adults with relapsed FLT3 mutant AML, and trials are underway to incorporate these agents into regimens for children [139]. Other mutations (eg, KIT, IDH1/2) that are amenable to therapeutic targeting are also being evaluated for children.

Therapeutic strategies designed to target AML at the molecular level, including epigenetic modifiers and targeted inhibitors, are active areas of investigation in pediatric AML [140]. Liposomal daunorubicin-cytarabine (CPX-351) has efficacy in adults with AML with unfavorable features (eg, therapy-related AML) and preliminary reports suggest efficacy in pediatric studies [141-143]. CD33 is expressed on the majority of pediatric patients with AML, and its high expression has also been associated with poor prognostic features [144-146]. Additional antibody-drug conjugates directed against CD33, as well as alternative antigens, such as CD123, and additional strategies including chimeric antigen receptor T cells (CAR-T), novel T cell engagers (eg, dual affinity targeting receptors), and natural killer (NK) cell therapy are currently being investigated in AML [147-153]. A number of clinical trials investigating these strategies are currently enrolling patients with relapsed and refractory AML, including children.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Acute myeloid leukemia".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient education" and the keyword(s) of interest.)

Basics topic (see "Patient education: Leukemia in children (The Basics)")

Basic topic (See "Patient education: Acute myeloid leukemia (AML) (The Basics)".)

SUMMARY AND RECOMMENDATIONS

Acute leukemia is the most common cancer in children. Acute lymphoblastic leukemia (ALL) accounts for the vast majority of pediatric acute leukemia. In contrast, acute myeloid leukemia (AML) is much less common in the pediatric population, representing approximately 15 percent. While survival rates have improved over the past several decades, survival in children with AML is lower than that of children with ALL.

Children with AML commonly present with fever, malaise, musculoskeletal pains, lymphadenopathy, hepatosplenomegaly, and bleeding. Blood work usually demonstrates anemia and thrombocytopenia. The white blood cell count can be decreased, normal, or increased. Less common complications that may require urgent intervention include disseminated intravascular coagulation (DIC), signs and symptoms related to central nervous system involvement, hyperleukocytosis, and tumor lysis syndrome. (See 'Clinical presentation' above.)

The diagnostic evaluation of children and adolescents with AML should include an evaluation for these potential complications along with a pathologic review of the morphology, immunophenotype, and genetic features. These studies both distinguish AML from ALL and other entities and provide information needed for risk stratification. (See 'Diagnostic evaluation' above and 'Pathologic features' above.)

Risk stratification incorporates cytogenetic and molecular features, genetic profiles, and response to induction chemotherapy. Treatment on research protocols is recommended as such protocols have helped to standardize treatment, improve survival rates, and decrease complications of therapy. Chemotherapeutic regimens in children and adolescents most often contain two courses of intensive induction chemotherapy followed by either cytarabine-based consolidation or allogeneic hematopoietic cell transplantation (HCT). Central nervous system prophylaxis is a standard component of therapy. (See 'Treatment' above.)

Patients undergoing AML therapy experience significant myelosuppression and have a high risk of infectious complications, but we suggest not routinely administering prophylactic antibiotics for children and adolescents receiving intensive chemotherapy for AML (Grade 2B). Fluoroquinolone prophylaxis can reduce the incidence of bacteremia and febrile neutropenia, but has not been shown to reduce sepsis or survival; these benefits must be balanced against the potential for toxicity, complications, and emergence of antibiotic resistance. Some experts do suggest routine prophylaxis with levofloxacin in this setting. (See 'Bacterial infections' above.)

New or prolonged fevers in the setting of neutropenia should prompt suspicion of fungal infection, and empiric antifungal therapy should be administered immediately for presumed or documented fungal infections. Specific practices for antifungal prophylaxis and treatment vary between centers, and consultation with Infectious Disease specialists should be considered in cases of presumed or documented infections. (See 'Fungal infections' above.)

Although cure rates for children and adolescents with AML have approached 70 percent, outcomes for children with adverse prognostic biologic features and refractory or relapsed disease remains poor. Novel therapies for the high-risk patients are needed. (See 'Relapsed leukemia' above and 'Risk stratification' above.)

Children and adolescents treated for AML should also be monitored closely for long-term complications of treatment regimens and require regular follow-up throughout their lives. (See 'Supportive care' above and 'Late effects' above.)

Children with Down syndrome (DS) have an increased risk for developing AML. AML in children with DS has unique biologic characteristics that greatly influence prognosis and therapy. (See 'Down syndrome and AML' above.)

  1. Gamis AS, Alonzo TA, Perentesis JP, et al. Children's Oncology Group's 2013 blueprint for research: acute myeloid leukemia. Pediatr Blood Cancer 2013; 60:964.
  2. Creutzig U, van den Heuvel-Eibrink MM, Gibson B, et al. Diagnosis and management of acute myeloid leukemia in children and adolescents: recommendations from an international expert panel. Blood 2012; 120:3187.
  3. Pui CH, Schrappe M, Ribeiro RC, Niemeyer CM. Childhood and adolescent lymphoid and myeloid leukemia. Hematology Am Soc Hematol Educ Program 2004; :118.
  4. Bolouri H, Farrar JE, Triche T Jr, et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat Med 2018; 24:103.
  5. Tarlock K, Zhong S, He Y, et al. Distinct age-associated molecular profiles in acute myeloid leukemia defined by comprehensive clinical genomic profiling. Oncotarget 2018; 9:26417.
  6. Gibson BE, Webb DK, Howman AJ, et al. Results of a randomized trial in children with Acute Myeloid Leukaemia: medical research council AML12 trial. Br J Haematol 2011; 155:366.
  7. Gamis AS, Alonzo TA, Meshinchi S, et al. Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531. J Clin Oncol 2014; 32:3021.
  8. Klein K, Kaspers G, Harrison CJ, et al. Clinical Impact of Additional Cytogenetic Aberrations, cKIT and RAS Mutations, and Treatment Elements in Pediatric t(8;21)-AML: Results From an International Retrospective Study by the International Berlin-Frankfurt-Münster Study Group. J Clin Oncol 2015; 33:4247.
  9. Lo-Coco F, Ammatuna E, Montesinos P, Sanz MA. Acute promyelocytic leukemia: recent advances in diagnosis and management. Semin Oncol 2008; 35:401.
  10. Tarlock K, Alonzo TA, Moraleda PP, et al. Acute myeloid leukaemia (AML) with t(6;9)(p23;q34) is associated with poor outcome in childhood AML regardless of FLT3-ITD status: a report from the Children's Oncology Group. Br J Haematol 2014; 166:254.
  11. Inaba H, Zhou Y, Abla O, et al. Heterogeneous cytogenetic subgroups and outcomes in childhood acute megakaryoblastic leukemia: a retrospective international study. Blood 2015; 126:1575.
  12. Balgobind BV, Zwaan CM, Pieters R, Van den Heuvel-Eibrink MM. The heterogeneity of pediatric MLL-rearranged acute myeloid leukemia. Leukemia 2011; 25:1239.
  13. Cooper TM, Ries RE, Alonzo TA, et al. Revised risk stratification criteria for children with newly diagnosed acute myeloid leukemia: a report from the Children’s Oncology Group. Blood 2017; 130:Abstract 407.
  14. Niktoreh N, Walter C, Zimmermann M, et al. Mutated WT1, FLT3-ITD, and NUP98-NSD1 Fusion in Various Combinations Define a Poor Prognostic Group in Pediatric Acute Myeloid Leukemia. J Oncol 2019; 2019:1609128.
  15. Struski S, Lagarde S, Bories P, et al. NUP98 is rearranged in 3.8% of pediatric AML forming a clinical and molecular homogenous group with a poor prognosis. Leukemia 2017; 31:565.
  16. Hollink IH, van den Heuvel-Eibrink MM, Arentsen-Peters ST, et al. NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern. Blood 2011; 118:3645.
  17. Creutzig U, Büchner T, Sauerland MC, et al. Significance of age in acute myeloid leukemia patients younger than 30 years: a common analysis of the pediatric trials AML-BFM 93/98 and the adult trials AMLCG 92/99 and AMLSG HD93/98A. Cancer 2008; 112:562.
  18. Meshinchi S, Woods WG, Stirewalt DL, et al. Prevalence and prognostic significance of Flt3 internal tandem duplication in pediatric acute myeloid leukemia. Blood 2001; 97:89.
  19. Zwaan CM, Meshinchi S, Radich JP, et al. FLT3 internal tandem duplication in 234 children with acute myeloid leukemia: prognostic significance and relation to cellular drug resistance. Blood 2003; 102:2387.
  20. Meshinchi S, Alonzo TA, Stirewalt DL, et al. Clinical implications of FLT3 mutations in pediatric AML. Blood 2006; 108:3654.
  21. Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood 2014; 124:3441.
  22. Ho PA, Alonzo TA, Gerbing RB, et al. Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): a report from the Children's Oncology Group. Blood 2009; 113:6558.
  23. Brown P, McIntyre E, Rau R, et al. The incidence and clinical significance of nucleophosmin mutations in childhood AML. Blood 2007; 110:979.
  24. Pollard JA, Alonzo TA, Gerbing RB, et al. Prevalence and prognostic significance of KIT mutations in pediatric patients with core binding factor AML enrolled on serial pediatric cooperative trials for de novo AML. Blood 2010; 115:2372.
  25. Ho PA, Zeng R, Alonzo TA, et al. Prevalence and prognostic implications of WT1 mutations in pediatric acute myeloid leukemia (AML): a report from the Children's Oncology Group. Blood 2010; 116:702.
  26. Hollink IH, van den Heuvel-Eibrink MM, Zimmermann M, et al. Clinical relevance of Wilms tumor 1 gene mutations in childhood acute myeloid leukemia. Blood 2009; 113:5951.
  27. Balgobind BV, Hollink IH, Arentsen-Peters ST, et al. Integrative analysis of type-I and type-II aberrations underscores the genetic heterogeneity of pediatric acute myeloid leukemia. Haematologica 2011; 96:1478.
  28. Vujkovic M, Attiyeh EF, Ries RE, et al. Genomic architecture and treatment outcome in pediatric acute myeloid leukemia: a Children's Oncology Group report. Blood 2017; 129:3051.
  29. Eidenschink Brodersen L, Alonzo TA, Menssen AJ, et al. A recurrent immunophenotype at diagnosis independently identifies high-risk pediatric acute myeloid leukemia: a report from Children's Oncology Group. Leukemia 2016; 30:2077.
  30. Gronseth CM, McElhone SE, Storer BE, et al. Prognostic significance of acquired copy-neutral loss of heterozygosity in acute myeloid leukemia. Cancer 2015; 121:2900.
  31. van der Velden VH, van der Sluijs-Geling A, Gibson BE, et al. Clinical significance of flowcytometric minimal residual disease detection in pediatric acute myeloid leukemia patients treated according to the DCOG ANLL97/MRC AML12 protocol. Leukemia 2010; 24:1599.
  32. Loken MR, Alonzo TA, Pardo L, et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children's Oncology Group. Blood 2012; 120:1581.
  33. Buldini B, Rizzati F, Masetti R, et al. Prognostic significance of flow-cytometry evaluation of minimal residual disease in children with acute myeloid leukaemia treated according to the AIEOP-AML 2002/01 study protocol. Br J Haematol 2017; 177:116.
  34. MRD-AML-BFM Study Group, Langebrake C, Creutzig U, et al. Residual disease monitoring in childhood acute myeloid leukemia by multiparameter flow cytometry: the MRD-AML-BFM Study Group. J Clin Oncol 2006; 24:3686.
  35. Steinbach D, Bader P, Willasch A, et al. Prospective validation of a new method of monitoring minimal residual disease in childhood acute myelogenous leukemia. Clin Cancer Res 2015; 21:1353.
  36. Rizzari C, Cazzaniga G, Coliva T, et al. Predictive factors of relapse and survival in childhood acute myeloid leukemia: role of minimal residual disease. Expert Rev Anticancer Ther 2011; 11:1391.
  37. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/209401s006lbl.pdf (Accessed on April 21, 2021).
  38. Stevens RF, Hann IM, Wheatley K, Gray RG. Marked improvements in outcome with chemotherapy alone in paediatric acute myeloid leukemia: results of the United Kingdom Medical Research Council's 10th AML trial. MRC Childhood Leukaemia Working Party. Br J Haematol 1998; 101:130.
  39. Gibson BE, Wheatley K, Hann IM, et al. Treatment strategy and long-term results in paediatric patients treated in consecutive UK AML trials. Leukemia 2005; 19:2130.
  40. Lie SO, Abrahamsson J, Clausen N, et al. Treatment stratification based on initial in vivo response in acute myeloid leukaemia in children without Down's syndrome: results of NOPHO-AML trials. Br J Haematol 2003; 122:217.
  41. Tsukimoto I, Tawa A, Horibe K, et al. Risk-stratified therapy and the intensive use of cytarabine improves the outcome in childhood acute myeloid leukemia: the AML99 trial from the Japanese Childhood AML Cooperative Study Group. J Clin Oncol 2009; 27:4007.
  42. Rihani R, Bazzeh F, Faqih N, Sultan I. Secondary hematopoietic malignancies in survivors of childhood cancer: an analysis of 111 cases from the Surveillance, Epidemiology, and End Result-9 registry. Cancer 2010; 116:4385.
  43. Woods WG, Neudorf S, Gold S, et al. A comparison of allogeneic bone marrow transplantation, autologous bone marrow transplantation, and aggressive chemotherapy in children with acute myeloid leukemia in remission. Blood 2001; 97:56.
  44. Ravindranath Y, Yeager AM, Chang MN, et al. Autologous bone marrow transplantation versus intensive consolidation chemotherapy for acute myeloid leukemia in childhood. Pediatric Oncology Group. N Engl J Med 1996; 334:1428.
  45. Oliansky DM, Rizzo JD, Aplan PD, et al. The role of cytotoxic therapy with hematopoietic stem cell transplantation in the therapy of acute myeloid leukemia in children: an evidence-based review. Biol Blood Marrow Transplant 2007; 13:1.
  46. Horan JT, Alonzo TA, Lyman GH, et al. Impact of disease risk on efficacy of matched related bone marrow transplantation for pediatric acute myeloid leukemia: the Children's Oncology Group. J Clin Oncol 2008; 26:5797.
  47. Kelly MJ, Horan JT, Alonzo TA, et al. Comparable survival for pediatric acute myeloid leukemia with poor-risk cytogenetics following chemotherapy, matched related donor, or unrelated donor transplantation. Pediatr Blood Cancer 2014; 61:269.
  48. Klusmann JH, Reinhardt D, Zimmermann M, et al. The role of matched sibling donor allogeneic stem cell transplantation in pediatric high-risk acute myeloid leukemia: results from the AML-BFM 98 study. Haematologica 2012; 97:21.
  49. Creutzig U, Zimmermann M, Ritter J, et al. Treatment strategies and long-term results in paediatric patients treated in four consecutive AML-BFM trials. Leukemia 2005; 19:2030.
  50. Lange BJ, Smith FO, Feusner J, et al. Outcomes in CCG-2961, a children's oncology group phase 3 trial for untreated pediatric acute myeloid leukemia: a report from the children's oncology group. Blood 2008; 111:1044.
  51. Niewerth D, Creutzig U, Bierings MB, Kaspers GJ. A review on allogeneic stem cell transplantation for newly diagnosed pediatric acute myeloid leukemia. Blood 2010; 116:2205.
  52. Meshinchi S, Arceci RJ, Sanders JE, et al. Role of allogeneic stem cell transplantation in FLT3/ITD-positive AML. Blood 2006; 108:400; author reply 400.
  53. Schechter T, Gassas A, Chen H, et al. The outcome of allogeneic hematopoietic cell transplantation for children with FMS-like tyrosine kinase 3 internal tandem duplication-positive acute myelogenous leukemia. Biol Blood Marrow Transplant 2015; 21:172.
  54. Moore J, Nivison-Smith I, Goh K, et al. Equivalent survival for sibling and unrelated donor allogeneic stem cell transplantation for acute myelogenous leukemia. Biol Blood Marrow Transplant 2007; 13:601.
  55. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med 2017; 377:454.
  56. Pollard JA, Alonzo TA, Brown PA, et al. Sorafenib in combination with standard chemotherapy for children with high allelic ratio FLT3/ITD+ AML improves event-free survival and reduces relapse risk: A Report from the Children’s Oncology Group Protocol AAML1031. Blood 2019; 135:Abstract 292.
  57. Chen YB, Li S, Lane AA, et al. Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia. Biol Blood Marrow Transplant 2014; 20:2042.
  58. Brunner AM, Li S, Fathi AT, et al. Haematopoietic cell transplantation with and without sorafenib maintenance for patients with FLT3-ITD acute myeloid leukaemia in first complete remission. Br J Haematol 2016; 175:496.
  59. Schlenk RF, Weber D, Fiedler W, et al. Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD. Blood 2019; 133:840.
  60. Tarlock K, Chang B, Cooper T, et al. Sorafenib treatment following hematopoietic stem cell transplant in pediatric FLT3/ITD acute myeloid leukemia. Pediatr Blood Cancer 2015; 62:1048.
  61. Dahl GV, Simone JV, Hustu HO, Mason C. Preventive central nervous system irradiation in children with acute nonlymphocytic leukemia. Cancer 1978; 42:2187.
  62. Chow EJ, Liu W, Srivastava K, et al. Differential effects of radiotherapy on growth and endocrine function among acute leukemia survivors: a childhood cancer survivor study report. Pediatr Blood Cancer 2013; 60:110.
  63. Kadan-Lottick NS, Zeltzer LK, Liu Q, et al. Neurocognitive functioning in adult survivors of childhood non-central nervous system cancers. J Natl Cancer Inst 2010; 102:881.
  64. Johnston DL, Alonzo TA, Gerbing RB, et al. Superior outcome of pediatric acute myeloid leukemia patients with orbital and CNS myeloid sarcoma: a report from the Children's Oncology Group. Pediatr Blood Cancer 2012; 58:519.
  65. Abbott BL, Rubnitz JE, Tong X, et al. Clinical significance of central nervous system involvement at diagnosis of pediatric acute myeloid leukemia: a single institution's experience. Leukemia 2003; 17:2090.
  66. Dusenbery KE, Howells WB, Arthur DC, et al. Extramedullary leukemia in children with newly diagnosed acute myeloid leukemia: a report from the Children's Cancer Group. J Pediatr Hematol Oncol 2003; 25:760.
  67. Shimizu H, Saitoh T, Hatsumi N, et al. Clinical significance of granulocytic sarcoma in adult patients with acute myeloid leukemia. Cancer Sci 2012; 103:1513.
  68. Hall MD, Chen YJ, Schultheiss TE, et al. Treatment outcomes for patients with chloroma receiving radiation therapy. J Med Imaging Radiat Oncol 2014; 58:523.
  69. Sanz MA, Lo Coco F, Martín G, et al. Definition of relapse risk and role of nonanthracycline drugs for consolidation in patients with acute promyelocytic leukemia: a joint study of the PETHEMA and GIMEMA cooperative groups. Blood 2000; 96:1247.
  70. Breen KA, Grimwade D, Hunt BJ. The pathogenesis and management of the coagulopathy of acute promyelocytic leukaemia. Br J Haematol 2012; 156:24.
  71. Petrie K, Zelent A, Waxman S. Differentiation therapy of acute myeloid leukemia: past, present and future. Curr Opin Hematol 2009; 16:84.
  72. Avvisati G, Lo-Coco F, Paoloni FP, et al. AIDA 0493 protocol for newly diagnosed acute promyelocytic leukemia: very long-term results and role of maintenance. Blood 2011; 117:4716.
  73. Powell BL, Moser B, Stock W, et al. Arsenic trioxide improves event-free and overall survival for adults with acute promyelocytic leukemia: North American Leukemia Intergroup Study C9710. Blood 2010; 116:3751.
  74. Abla O, Ribeiro RC. How I treat children and adolescents with acute promyelocytic leukaemia. Br J Haematol 2014; 164:24.
  75. Testi AM, Pession A, Diverio D, et al. Risk-adapted treatment of acute promyelocytic leukemia: results from the International Consortium for Childhood APL. Blood 2018; 132:405.
  76. Lo-Coco F, Avvisati G, Vignetti M, et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med 2013; 369:111.
  77. Platzbecker U, Avvisati G, Cicconi L, et al. Improved Outcomes With Retinoic Acid and Arsenic Trioxide Compared With Retinoic Acid and Chemotherapy in Non-High-Risk Acute Promyelocytic Leukemia: Final Results of the Randomized Italian-German APL0406 Trial. J Clin Oncol 2017; 35:605.
  78. Garcia Spezza E, Brethon B, Petit A, et al. Tolerance to arsenic trioxide combined with all-trans-retinoic acid in children with acute promyelocytic leukaemia in France. Br J Haematol 2020; 188:170.
  79. Klusmann JH, Creutzig U, Zimmermann M, et al. Treatment and prognostic impact of transient leukemia in neonates with Down syndrome. Blood 2008; 111:2991.
  80. Al-Kasim F, Doyle JJ, Massey GV, et al. Incidence and treatment of potentially lethal diseases in transient leukemia of Down syndrome: Pediatric Oncology Group Study. J Pediatr Hematol Oncol 2002; 24:9.
  81. Gamis AS, Alonzo TA, Gerbing RB, et al. Natural history of transient myeloproliferative disorder clinically diagnosed in Down syndrome neonates: a report from the Children's Oncology Group Study A2971. Blood 2011; 118:6752.
  82. Hama A, Yagasaki H, Takahashi Y, et al. Acute megakaryoblastic leukaemia (AMKL) in children: a comparison of AMKL with and without Down syndrome. Br J Haematol 2008; 140:552.
  83. Lange BJ, Kobrinsky N, Barnard DR, et al. Distinctive demography, biology, and outcome of acute myeloid leukemia and myelodysplastic syndrome in children with Down syndrome: Children's Cancer Group Studies 2861 and 2891. Blood 1998; 91:608.
  84. Wechsler J, Greene M, McDevitt MA, et al. Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome. Nat Genet 2002; 32:148.
  85. Hitzler JK, Cheung J, Li Y, et al. GATA1 mutations in transient leukemia and acute megakaryoblastic leukemia of Down syndrome. Blood 2003; 101:4301.
  86. Taub JW, Huang X, Matherly LH, et al. Expression of chromosome 21-localized genes in acute myeloid leukemia: differences between Down syndrome and non-Down syndrome blast cells and relationship to in vitro sensitivity to cytosine arabinoside and daunorubicin. Blood 1999; 94:1393.
  87. Ge Y, Jensen TL, Stout ML, et al. The role of cytidine deaminase and GATA1 mutations in the increased cytosine arabinoside sensitivity of Down syndrome myeloblasts and leukemia cell lines. Cancer Res 2004; 64:728.
  88. Ravindranath Y, Abella E, Krischer JP, et al. Acute myeloid leukemia (AML) in Down's syndrome is highly responsive to chemotherapy: experience on Pediatric Oncology Group AML Study 8498. Blood 1992; 80:2210.
  89. Creutzig U, Reinhardt D, Diekamp S, et al. AML patients with Down syndrome have a high cure rate with AML-BFM therapy with reduced dose intensity. Leukemia 2005; 19:1355.
  90. Zeller B, Gustafsson G, Forestier E, et al. Acute leukaemia in children with Down syndrome: a population-based Nordic study. Br J Haematol 2005; 128:797.
  91. Sorrell AD, Alonzo TA, Hilden JM, et al. Favorable survival maintained in children who have myeloid leukemia associated with Down syndrome using reduced-dose chemotherapy on Children's Oncology Group trial A2971: a report from the Children's Oncology Group. Cancer 2012; 118:4806.
  92. Taub JW, Berman JN, Hitzler JK, et al. Improved outcomes for myeloid leukemia of Down syndrome: a report from the Children's Oncology Group AAML0431 trial. Blood 2017; 129:3304.
  93. O'Brien MM, Taub JW, Chang MN, et al. Cardiomyopathy in children with Down syndrome treated for acute myeloid leukemia: a report from the Children's Oncology Group Study POG 9421. J Clin Oncol 2008; 26:414.
  94. Lie SO, Jonmundsson G, Mellander L, et al. A population-based study of 272 children with acute myeloid leukaemia treated on two consecutive protocols with different intensity: best outcome in girls, infants, and children with Down's syndrome. Nordic Society of Paediatric Haematology and Oncology (NOPHO). Br J Haematol 1996; 94:82.
  95. Kojima S, Sako M, Kato K, et al. An effective chemotherapeutic regimen for acute myeloid leukemia and myelodysplastic syndrome in children with Down's syndrome. Leukemia 2000; 14:786.
  96. Uffmann M, Rasche M, Zimmermann M, et al. Therapy reduction in patients with Down syndrome and myeloid leukemia: the international ML-DS 2006 trial. Blood 2017; 129:3314.
  97. Muramatsu H, Sakaguchi H, Taga T, et al. Reduced intensity conditioning in allogeneic stem cell transplantation for AML with Down syndrome. Pediatr Blood Cancer 2014; 61:925.
  98. Hitzler JK, He W, Doyle J, et al. Outcome of transplantation for acute myelogenous leukemia in children with Down syndrome. Biol Blood Marrow Transplant 2013; 19:893.
  99. Creutzig U, Ritter J, Budde M, et al. Early deaths due to hemorrhage and leukostasis in childhood acute myelogenous leukemia. Associations with hyperleukocytosis and acute monocytic leukemia. Cancer 1987; 60:3071.
  100. Bunin NJ, Pui CH. Differing complications of hyperleukocytosis in children with acute lymphoblastic or acute nonlymphoblastic leukemia. J Clin Oncol 1985; 3:1590.
  101. von Neuhoff C, Reinhardt D, Sander A, et al. Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol 2010; 28:2682.
  102. Ganzel C, Becker J, Mintz PD, et al. Hyperleukocytosis, leukostasis and leukapheresis: practice management. Blood Rev 2012; 26:117.
  103. Porcu P, Farag S, Marcucci G, et al. Leukocytoreduction for acute leukemia. Ther Apher 2002; 6:15.
  104. Röllig C, Ehninger G. How I treat hyperleukocytosis in acute myeloid leukemia. Blood 2015; 125:3246.
  105. Oberoi S, Lehrnbecher T, Phillips B, et al. Leukapheresis and low-dose chemotherapy do not reduce early mortality in acute myeloid leukemia hyperleukocytosis: a systematic review and meta-analysis. Leuk Res 2014; 38:460.
  106. Canner J, Alonzo TA, Franklin J, et al. Differences in outcomes of newly diagnosed acute myeloid leukemia for adolescent/young adult and younger patients: a report from the Children's Oncology Group. Cancer 2013; 119:4162.
  107. Rubnitz JE, Pounds S, Cao X, et al. Treatment outcome in older patients with childhood acute myeloid leukemia. Cancer 2012; 118:6253.
  108. Alexander S, Fisher BT, Gaur AH, et al. Effect of Levofloxacin Prophylaxis on Bacteremia in Children With Acute Leukemia or Undergoing Hematopoietic Stem Cell Transplantation: A Randomized Clinical Trial. JAMA 2018; 320:995.
  109. Creutzig U, Zimmermann M, Lehrnbecher T, et al. Less toxicity by optimizing chemotherapy, but not by addition of granulocyte colony-stimulating factor in children and adolescents with acute myeloid leukemia: results of AML-BFM 98. J Clin Oncol 2006; 24:4499.
  110. Lehrnbecher T, Sung L. Anti-infective prophylaxis in pediatric patients with acute myeloid leukemia. Expert Rev Hematol 2014; 7:819.
  111. Feusner JH, Hastings CA. Infections in children with acute myelogenous leukemia. Concepts of management and prevention. J Pediatr Hematol Oncol 1995; 17:234.
  112. Lewis V, Yanofsky R, Mitchell D, et al. Predictors and outcomes of viridans group streptococcal infections in pediatric acute myeloid leukemia: from the Canadian infections in AML research group. Pediatr Infect Dis J 2014; 33:126.
  113. Okamoto Y, Ribeiro RC, Srivastava DK, et al. Viridans streptococcal sepsis: clinical features and complications in childhood acute myeloid leukemia. J Pediatr Hematol Oncol 2003; 25:696.
  114. Creutzig U, Zimmermann M, Reinhardt D, et al. Early deaths and treatment-related mortality in children undergoing therapy for acute myeloid leukemia: analysis of the multicenter clinical trials AML-BFM 93 and AML-BFM 98. J Clin Oncol 2004; 22:4384.
  115. Fisher BT, Kavcic M, Li Y, et al. Antifungal prophylaxis associated with decreased induction mortality rates and resources utilized in children with new-onset acute myeloid leukemia. Clin Infect Dis 2014; 58:502.
  116. Yamaguchi M, Kurokawa T, Ishiyama K, et al. Efficacy and safety of micafungin as an empirical therapy for invasive fungal infections in patients with hematologic disorders: a multicenter, prospective study. Ann Hematol 2011; 90:1209.
  117. Emiroglu M. Micafungin use in children. Expert Rev Anti Infect Ther 2011; 9:821.
  118. Patterson TF, Thompson GR 3rd, Denning DW, et al. Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis 2016; 63:e1.
  119. Rotstein C, Bow EJ, Laverdiere M, et al. Randomized placebo-controlled trial of fluconazole prophylaxis for neutropenic cancer patients: benefit based on purpose and intensity of cytotoxic therapy. The Canadian Fluconazole Prophylaxis Study Group. Clin Infect Dis 1999; 28:331.
  120. Mehta AK, Langston AA. Use of posaconazole in the treatment of invasive fungal  infections. Expert Rev Hematol 2009; 2:619.
  121. Cornely OA, Maertens J, Winston DJ, et al. Posaconazole vs. fluconazole or itraconazole prophylaxis in patients with neutropenia. N Engl J Med 2007; 356:348.
  122. Pagano L, Caira M, Candoni A, et al. Evaluation of the practice of antifungal prophylaxis use in patients with newly diagnosed acute myeloid leukemia: results from the SEIFEM 2010-B registry. Clin Infect Dis 2012; 55:1515.
  123. Glasmacher A, Prentice A, Gorschlüter M, et al. Itraconazole prevents invasive fungal infections in neutropenic patients treated for hematologic malignancies: evidence from a meta-analysis of 3,597 patients. J Clin Oncol 2003; 21:4615.
  124. Barton CD, Waugh LK, Nielsen MJ, Paulus S. Febrile neutropenia in children treated for malignancy. J Infect 2015; 71 Suppl 1:S27.
  125. Sander A, Zimmermann M, Dworzak M, et al. Consequent and intensified relapse therapy improved survival in pediatric AML: results of relapse treatment in 379 patients of three consecutive AML-BFM trials. Leukemia 2010; 24:1422.
  126. Cooper TM, Alonzo TA, Gerbing RB, et al. AAML0523: a report from the Children's Oncology Group on the efficacy of clofarabine in combination with cytarabine in pediatric patients with recurrent acute myeloid leukemia. Cancer 2014; 120:2482.
  127. Fleischhack G, Hasan C, Graf N, et al. IDA-FLAG (idarubicin, fludarabine, cytarabine, G-CSF), an effective remission-induction therapy for poor-prognosis AML of childhood prior to allogeneic or autologous bone marrow transplantation: experiences of a phase II trial. Br J Haematol 1998; 102:647.
  128. Kaspers GJ, Zimmermann M, Reinhardt D, et al. Improved outcome in pediatric relapsed acute myeloid leukemia: results of a randomized trial on liposomal daunorubicin by the International BFM Study Group. J Clin Oncol 2013; 31:599.
  129. Niktoreh N, Lerius B, Zimmermann M, et al. Gemtuzumab ozogamicin in children with relapsed or refractory acute myeloid leukemia: a report by Berlin-Frankfurt-Münster study group. Haematologica 2019; 104:120.
  130. Arceci RJ, Sande J, Lange B, et al. Safety and efficacy of gemtuzumab ozogamicin in pediatric patients with advanced CD33+ acute myeloid leukemia. Blood 2005; 106:1183.
  131. Lipshultz SE, Franco VI, Miller TL, et al. Cardiovascular disease in adult survivors of childhood cancer. Annu Rev Med 2015; 66:161.
  132. Kremer LC, van Dalen EC, Offringa M, et al. Anthracycline-induced clinical heart failure in a cohort of 607 children: long-term follow-up study. J Clin Oncol 2001; 19:191.
  133. Creutzig U, Diekamp S, Zimmermann M, Reinhardt D. Longitudinal evaluation of early and late anthracycline cardiotoxicity in children with AML. Pediatr Blood Cancer 2007; 48:651.
  134. Getz KD, Sung L, Ky B, et al. Occurrence of Treatment-Related Cardiotoxicity and Its Impact on Outcomes Among Children Treated in the AAML0531 Clinical Trial: A Report From the Children's Oncology Group. J Clin Oncol 2019; 37:12.
  135. Mulrooney DA, Dover DC, Li S, et al. Twenty years of follow-up among survivors of childhood and young adult acute myeloid leukemia: a report from the Childhood Cancer Survivor Study. Cancer 2008; 112:2071.
  136. Meadows AT, Friedman DL, Neglia JP, et al. Second neoplasms in survivors of childhood cancer: findings from the Childhood Cancer Survivor Study cohort. J Clin Oncol 2009; 27:2356.
  137. Park J, Choi EK, Kim JH, et al. Effects of total body irradiation-based conditioning on allogeneic stem cell transplantation for pediatric acute leukemia: a single-institution study. Radiat Oncol J 2014; 32:198.
  138. Chemaitilly W, Sklar CA. Endocrine complications in long-term survivors of childhood cancers. Endocr Relat Cancer 2010; 17:R141.
  139. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N Engl J Med 2019; 381:1728.
  140. Tasian SK, Pollard JA, Aplenc R. Molecular therapeutic approaches for pediatric acute myeloid leukemia. Front Oncol 2014; 4:55.
  141. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia. J Clin Oncol 2018; 36:2684.
  142. Cooper TM, Absalon M, Alonzo TA, et al. AAML1421, a phase I/II study of CPX-351 followed by fludarabine, cytarabine, and G-CSF (FLAG) for children with relapsed acute myeloid leukemia (AML): A report from the Children’s Oncology Group. J Clin Oncol 2019; 37:Abstract 10003.
  143. Absalon M, O'Brien MM, Phillips CL, et al. A phase I/pilot study of CPX-351 for children, adolescents and young adults with recurrent or refractory hematologic malignancies. J Clin Oncol 2016; 34:Abstract 10541.
  144. Pollard JA, Alonzo TA, Loken M, et al. Correlation of CD33 expression level with disease characteristics and response to gemtuzumab ozogamicin containing chemotherapy in childhood AML. Blood 2012; 119:3705.
  145. Walter RB, Gooley TA, van der Velden VH, et al. CD33 expression and P-glycoprotein-mediated drug efflux inversely correlate and predict clinical outcome in patients with acute myeloid leukemia treated with gemtuzumab ozogamicin monotherapy. Blood 2007; 109:4168.
  146. Sievers EL, Larson RA, Stadtmauer EA, et al. Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. J Clin Oncol 2001; 19:3244.
  147. Kung Sutherland MS, Walter RB, Jeffrey SC, et al. SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. Blood 2013; 122:1455.
  148. Krupka C, Kufer P, Kischel R, et al. CD33 target validation and sustained depletion of AML blasts in long-term cultures by the bispecific T-cell-engaging antibody AMG 330. Blood 2014; 123:356.
  149. Chichili GR, Huang L, Li H, et al. A CD3xCD123 bispecific DART for redirecting host T cells to myelogenous leukemia: preclinical activity and safety in nonhuman primates. Sci Transl Med 2015; 7:289ra82.
  150. He SZ, Busfield S, Ritchie DS, et al. A Phase 1 study of the safety, pharmacokinetics and anti-leukemic activity of the anti-CD123 monoclonal antibody CSL360 in relapsed, refractory or high-risk acute myeloid leukemia. Leuk Lymphoma 2015; 56:1406.
  151. Wang QS, Wang Y, Lv HY, et al. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther 2015; 23:184.
  152. Nguyen R, Wu H, Pounds S, et al. A phase II clinical trial of adoptive transfer of haploidentical natural killer cells for consolidation therapy of pediatric acute myeloid leukemia. J Immunother Cancer 2019; 7:81.
  153. Björklund AT, Carlsten M, Sohlberg E, et al. Complete Remission with Reduction of High-Risk Clones following Haploidentical NK-Cell Therapy against MDS and AML. Clin Cancer Res 2018; 24:1834.
Topic 13935 Version 25.0

References